Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
Gut Microbes ; 11(5): 1423-1437, 2020 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-32403971

RESUMO

The protein translocated intimin receptor (Tir) from enteropathogenic Escherichia coli shares sequence similarity with the host cellular immunoreceptor tyrosine-based inhibition motifs (ITIMs). The ITIMs of Tir are required for Tir-mediated immune inhibition and evasion of host immune responses. However, the underlying molecular mechanism by which Tir regulates immune inhibition remains unclear. Here we demonstrated that ß-arrestin 2, which is involved in the G-protein-coupled receptor (GPCR) signal pathway, interacted with Tir in an ITIM-dependent manner. For the molecular mechanism, we found that ß-arrestin 2 enhanced the recruitment of SHP-1 to Tir. The recruited SHP-1 inhibited K63-linked ubiquitination of TRAF6 by dephosphorylating TRAF6 at Tyr288, and inhibited K63-linked ubiquitination and phosphorylation of TAK1 by dephosphorylating TAK1 at Tyr206, which cut off the downstream signal transduction and subsequent cytokine production. Moreover, the inhibitory effect of Tir on immune responses was diminished in ß-arrestin 2-deficient mice and macrophages. These findings suggest that ß-arrestin 2 is a key regulator in Tir-mediated immune evasion, which could serve as a new therapeutic target for bacterial infectious diseases.


Assuntos
Escherichia coli Enteropatogênica/patogenicidade , Evasão da Resposta Imune , Macrófagos/microbiologia , Receptores Toll-Like/metabolismo , beta-Arrestina 2/metabolismo , Motivos de Aminoácidos , Animais , Células Cultivadas , Citocinas/genética , Citocinas/metabolismo , Escherichia coli Enteropatogênica/imunologia , Escherichia coli Enteropatogênica/metabolismo , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/metabolismo , Células HEK293 , Células HeLa , Humanos , MAP Quinase Quinase Quinases/metabolismo , Macrófagos Peritoneais/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação , Ligação Proteica , Proteína Tirosina Fosfatase não Receptora Tipo 6/metabolismo , Células RAW 264.7 , RNA Interferente Pequeno , Receptores de Superfície Celular/química , Receptores de Superfície Celular/metabolismo , Transdução de Sinais , Fator 6 Associado a Receptor de TNF/genética , Fator 6 Associado a Receptor de TNF/metabolismo , beta-Arrestina 2/genética
2.
J. pediatr. (Rio J.) ; 93(6): 568-575, Nov.-Dec. 2017. tab, graf
Artigo em Inglês | LILACS | ID: biblio-894078

RESUMO

Abstract Objective: Intimins are protein adhesins of enteropathogenic Escherichia coli and enterohemorrhagic E. coli capable of inducing attachment and effacement lesions in enterocytes. Anti-intimin antibodies are important for the protection from enteropathogenic E. coli and enterohemorrhagic E. coli infections because these antibodies inhibit bacterial adhesion and impair the initial step of the pathogenesis. We studied the transfer of maternal anti-intimin antibodies from healthy Brazilian mothers to their newborns through the placenta and colostrum. Methods: Serum immunoglobulin G and secretory immunoglobulin A antibodies against conserved and variable regions of intimins α, β, and γ were analyzed using an enzyme linked-immunosorbent assay in the blood and colostrum from 45 healthy women as well as cord blood serum samples from their newborns. Results: The concentrations of antibodies reactive with α intimin were significantly lower than those of anti-γ and anti-conserved intimin antibodies in the colostrum samples. IgG serum antibodies reactive with all the subtypes of intimins were transferred to the newborns, but the concentrations of anti-conserved intimin serum antibodies were significantly higher in mothers and newborns than concentrations of antibodies against variable regions. The patterns of IgG transfer from mothers to newborns were similar for all anti-intimin antibodies. These values are similar to the percentage transference of total IgG. Conclusions: Anti-intimin antibodies are transferred from mothers to newborns through the placenta, and reinforce the protection provided by breastfeeding against diarrheagenic E. coli infections.


Resumo Objetivo: As intiminas são adesinas proteicas de Escherichia coli enteropatogênicas (EPEC) e enterro-hemorrágicas (EHEC) capazes de induzir as lesões attaching and effacing nos enterócitos. Anticorpos anti-intiminas são importantes para a proteção contra infecções por EPEC e EHEC porque esses anticorpos inibem a adesão bacteriana e impedem o passo inicial do mecanismo patogênico dessas bactérias. Nós estudamos a transferência de anticorpos maternos anti-intiminas de mães brasileiras saudáveis para os seus recém-nascidos através da placenta e do colostro. Métodos: Anticorpos séricos da classe IgG e secretórios da classe IgA (SIgA) reativos com as porções conservada (cons) e variáveis das intiminas α (vα), β (vβ) e γ (vγ) foram analisados pelo teste de ELISA no sangue e no colostro de 45 parturientes saudáveis e no sangue de cordão umbilical dos seus respectivos recém-nascidos. Resultados: As concentrações de anticorpos reativos com intimina vα foram significativamente mais baixas que as dos anticorpos anti-vγ e anti-cons nas amostras de colostro. Anticorpos IgG séricos reativos com todas as intiminas foram transferidos para os recém-nascidos, mas as concentrações de anti-cons foram significativamente mais altas tanto nas mães como nos recém-nascidos do que os anticorpos reativos com as regiões variáveis das intiminas. O padrão de transferência de IgG das mães para os recém-nascidos foi muito semelhante para todos os anticorpos anti-intiminas. Os valores de porcentagem de transferência foram semelhantes à transferência de IgG total. Conclusões: Anticorpos anti-intimina são transferidos das mães para os recém-nascidos pela placenta e corroboram a proteção contra infecções por Escherichia coli diarreiogênicas (DEC) conferida pelo aleitamento materno.


Assuntos
Humanos , Feminino , Recém-Nascido , Autoanticorpos/análise , Imunoglobulina A Secretora/análise , Imunoglobulina G/análise , Colostro/imunologia , Escherichia coli Enteropatogênica/imunologia , Sangue Fetal/imunologia , Ensaio de Imunoadsorção Enzimática , Adesinas Bacterianas/análise , Adesinas Bacterianas/imunologia , Proteínas de Escherichia coli/análise , Proteínas de Escherichia coli/imunologia
4.
Nat Commun ; 8(1): 1576, 2017 11 17.
Artigo em Inglês | MEDLINE | ID: mdl-29146903

RESUMO

Dysregulation of autophagy and inflammasome activity contributes to the development of auto-inflammatory diseases. Emerging evidence highlights the importance of the actin cytoskeleton in modulating inflammatory responses. Here we show that deficiency of Wiskott-Aldrich syndrome protein (WASp), which signals to the actin cytoskeleton, modulates autophagy and inflammasome function. In a model of sterile inflammation utilizing TLR4 ligation followed by ATP or nigericin treatment, inflammasome activation is enhanced in monocytes from WAS patients and in WAS-knockout mouse dendritic cells. In ex vivo models of enteropathogenic Escherichia coli and Shigella flexneri infection, WASp deficiency causes defective bacterial clearance, excessive inflammasome activation and host cell death that are associated with dysregulated septin cage-like formation, impaired autophagic p62/LC3 recruitment and defective formation of canonical autophagosomes. Taken together, we propose that dysregulation of autophagy and inflammasome activities contribute to the autoinflammatory manifestations of WAS, thereby identifying potential targets for therapeutic intervention.


Assuntos
Citoesqueleto de Actina/metabolismo , Autofagia/imunologia , Inflamassomos/imunologia , Proteína da Síndrome de Wiskott-Aldrich/genética , Proteína da Síndrome de Wiskott-Aldrich/metabolismo , Síndrome de Wiskott-Aldrich/imunologia , Animais , Autofagia/genética , Carga Bacteriana/imunologia , Linhagem Celular Tumoral , Células Dendríticas/imunologia , Escherichia coli Enteropatogênica/imunologia , Humanos , Imunidade Inata/imunologia , Interferon Tipo I/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/imunologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/imunologia , Nigericina/farmacologia , Septinas/metabolismo , Shigella flexneri/imunologia , Células THP-1 , Receptor 4 Toll-Like/imunologia , Síndrome de Wiskott-Aldrich/metabolismo
5.
PLoS Pathog ; 11(9): e1005121, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26332984

RESUMO

Enteropathogenic and enterohemorrhagic Escherichia coli (EPEC and EHEC) are related strains capable of inducing severe gastrointestinal disease. For optimal infection, these pathogens actively modulate cellular functions through the deployment of effector proteins in a type three secretion system (T3SS)-dependent manner. In response to enteric pathogen invasion, the Nod-like receptor pyrin domain containing (NLRP) inflammasome has been increasingly recognized as an important cytoplasmic sensor against microbial infection by activating caspase-1 and releasing IL-1ß. EPEC and EHEC are known to elicit inflammasome activation in macrophages and epithelial cells; however, whether the pathogens actively counteract such innate immune responses is unknown. Using a series of compound effector-gene deletion strains of EPEC, we screened and identified NleA, which could subdue host IL-1ß secretion. It was found that the reduction is not because of blocked NF-κB activity; instead, the reduction results from inhibited caspase-1 activation by NleA. Immunostaining of human macrophage-like cells following infection revealed limited formation of inflammasome foci with constituents of total caspase-1, ASC and NLRP3 in the presence of NleA. Pulldown of PMA-induced differentiated THP-1 lysate with purified MBP-NleA reveals that NLRP3 is a target of NleA. The interaction was verified by an immunoprecipitation assay and direct interaction assay in which purified MBP-NleA and GST-NLRP3 were used. We further showed that the effector interacts with regions of NLRP3 containing the PYD and LRR domains. Additionally, NleA was found to associate with non-ubiquitinated and ubiquitinated NLRP3 and to interrupt de-ubiquitination of NLRP3, which is a required process for inflammasome activation. Cumulatively, our findings provide the first example of EPEC-mediated suppression of inflammasome activity in which NieA plays a novel role in controlling the host immune response through targeting of NLRP3.


Assuntos
Proteínas de Transporte/antagonistas & inibidores , Escherichia coli Enteropatogênica/fisiologia , Infecções por Escherichia coli/microbiologia , Proteínas de Escherichia coli/metabolismo , Interações Hospedeiro-Patógeno , Inflamassomos/metabolismo , Macrófagos/microbiologia , Fatores de Virulência/metabolismo , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Caspase 1/química , Caspase 1/metabolismo , Linhagem Celular , Escherichia coli Enteropatogênica/imunologia , Ativação Enzimática , Infecções por Escherichia coli/imunologia , Infecções por Escherichia coli/metabolismo , Infecções por Escherichia coli/patologia , Proteínas de Escherichia coli/genética , Deleção de Genes , Células HeLa , Humanos , Imunidade Inata , Inflamassomos/imunologia , Interleucina-1beta/antagonistas & inibidores , Interleucina-1beta/metabolismo , Cinética , Proteínas Luminescentes/química , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Macrófagos/imunologia , Macrófagos/metabolismo , Macrófagos/patologia , Proteína 3 que Contém Domínio de Pirina da Família NLR , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/genética , Fragmentos de Peptídeos/metabolismo , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Ubiquitinação , Fatores de Virulência/genética
6.
Cell Host Microbe ; 16(2): 249-256, 2014 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-25121752

RESUMO

Inflammasome-mediated host defenses have been extensively studied in innate immune cells. Whether inflammasomes function for innate defense in intestinal epithelial cells, which represent the first line of defense against enteric pathogens, remains unknown. We observed enhanced Salmonella enterica serovar Typhimurium colonization in the intestinal epithelium of caspase-11-deficient mice, but not at systemic sites. In polarized epithelial monolayers, siRNA-mediated depletion of caspase-4, a human ortholog of caspase-11, also led to increased bacterial colonization. Decreased rates of pyroptotic cell death, a host defense mechanism that extrudes S. Typhimurium-infected cells from the polarized epithelium, accounted for increased pathogen burdens. The caspase-4 inflammasome also governs activation of the proinflammatory cytokine, interleukin (IL)-18, in response to intracellular (S. Typhimurium) and extracellular (enteropathogenic Escherichia coli) enteric pathogens, via intracellular LPS sensing. Therefore, an epithelial cell-intrinsic noncanonical inflammasome plays a critical role in antimicrobial defense at the intestinal mucosal surface.


Assuntos
Caspases Iniciadoras/metabolismo , Caspases/metabolismo , Infecções por Escherichia coli/enzimologia , Inflamassomos/fisiologia , Infecções por Salmonella/enzimologia , Animais , Linhagem Celular Tumoral , Escherichia coli Enteropatogênica/imunologia , Ativação Enzimática , Infecções por Escherichia coli/imunologia , Gastroenterite/enzimologia , Gastroenterite/microbiologia , Humanos , Interleucina-18/metabolismo , Mucosa Intestinal/enzimologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Lipopolissacarídeos/farmacologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Infecções por Salmonella/imunologia , Salmonella enterica/imunologia
7.
J Microbiol Immunol Infect ; 47(2): 81-6, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23079484

RESUMO

BACKGROUND: Enteropathogenic Escherichia coli (EPEC) is an important cause of diarrhea in human. This study was conducted to investigate the ability of orally administrated probiotic lactobacilli to protect hosts from EPEC infection via enhancement of immune responses. METHODS: Lyophilized Lactobacillus gasseri TMC0356 (TMC0356) was orally administered to Institute of Cancer Research (ICR) mice and Sprague Dawley (SD) rats for 11 and 7 days, respectively. These tested mice and rats were intraperitoneally injected with EPEC. Body weight, general symptoms (piloerection, soft stool, diarrhea, and anal hyperemia), and mortality of the tested mice were observed. Peritoneal macrophages were extracted from peritoneal cavity of tested rats, and their phagocytosis and cytokine production were analyzed. RESULTS: Oral administration of TMC0356 accelerated the disappearance of general symptoms and reduced mortality of EPEC-infected mice in the early phase. Peritoneal macrophages from rats orally administered with TMC0356 showed significant increases in phagocytic activity (p < 0.05) and interleukin (IL)-6 production (p < 0.01) compared to those from control rats. Tumor necrosis factor-α and production of IL-1ß, IL-10, and IL-12 slightly increased, although the changes were not statistically significant. CONCLUSION: These results suggest that some of selected probiotic lactobacilli may, at least partly, protect hosts from EPEC infection by the enhancement of innate immunity of host and attenuate symptoms caused by the infection.


Assuntos
Escherichia coli Enteropatogênica/imunologia , Infecções por Escherichia coli/patologia , Fatores Imunológicos/administração & dosagem , Lactobacillus/imunologia , Macrófagos Peritoneais/imunologia , Peritonite/patologia , Probióticos/administração & dosagem , Administração Oral , Animais , Peso Corporal , Modelos Animais de Doenças , Infecções por Escherichia coli/imunologia , Masculino , Camundongos Endogâmicos ICR , Peritonite/imunologia , Ratos Sprague-Dawley , Resultado do Tratamento
8.
Microb Pathog ; 64: 48-56, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24036180

RESUMO

Enteropathogenic Escherichia coli (EPEC) uses a type III secretion system (T3SS) to inject effectors into host cells and alter cellular physiology. The aim of the present study was to identify targets of human secretory immunoglobulin A (sIgA) antibodies from the proteins delivered by EPEC into HEp-2 cells after infection. Bacterial proteins delivered into EPEC-infected cells were obtained in sub-cellular fractions (cytoplasmic, membrane, and cytoskeleton) and probed with sIgA antibodies from human milk and analyzed by Western blotting. These sIgA antibodies reacted with Tir and EspB in the cytoplasmic and membrane fractions, and with intimin in the membrane fraction mainly. The sIgA also identified an EPEC surface-associated Heat-shock protein 70 (Hsp70) in HEp-2 cells infected with EPEC. Purified Hsp70 from EPEC was able to bind to HEp-2 cells, suggesting adhesive properties in this protein. EspC secreted to the medium reacted strongly with the sIgA antibodies. An EPEC 115 kDa protein, unrelated to the EspC protein, was detected in the cytoplasm of infected HEp-2 cells, suggesting that this is a new protein translocated by EPEC. The results suggest that there is a strong host antibody response to Tir and intimin, which are essential proteins for attaching and effacing (A/E) pathogen mediated disease.


Assuntos
Escherichia coli Enteropatogênica/imunologia , Proteínas de Escherichia coli/imunologia , Imunoglobulina A Secretora/imunologia , Leite Humano/imunologia , Células Hep G2 , Humanos , Fatores de Virulência/imunologia
9.
Braz. j. microbiol ; 44(2): 499-504, 2013. tab
Artigo em Inglês | LILACS | ID: lil-688585

RESUMO

The therapeutic action of phosphorylated mannanoligosaccharides (MOS) was investigated regarding its prebiotic activity on enteropathogenic Escherichia coli (EPEC). Diarrhea was induced in dogs by experimental infection with EPEC strains. Then MOS was supplied once a day, in water for 20 days. Immunological (IgA and IgG), hematological (lymphocytes, neutrophils and monocytes) and bacteriological variables (PCR detection of the eae gene of EPEC recovered from stool culture), as well as occurrence of diarrhea were evaluated. All strains caused diarrhea at 24, 48 and 72 h after infection. PCR results indicated that E. coli isolated from stool culture of all infected animals had the eae gene. There was no significant difference among groups as to number of blood cells in the hemogram and IgA and IgG production. MOS was effective in recovering of EPEC-infected dogs since prebiotic-treated animals recovered more rapidly from infection than untreated ones (p < 0.05). This is an important finding since diarrhea causes intense dehydration and nutrient loss. The use of prebiotics for humans and other animals with diarrhea can be an alternative for the treatment and prophylaxis of EPEC infections.


Assuntos
Animais , Cães , Sangue/imunologia , Diarreia/microbiologia , Escherichia coli Enteropatogênica/imunologia , Fezes , Fármacos Gastrointestinais/metabolismo , Oligossacarídeos/metabolismo , Prebióticos , Anticorpos Antibacterianos/sangue , Fenômenos Químicos , Modelos Animais de Doenças , Escherichia coli , Fármacos Gastrointestinais/administração & dosagem , Fármacos Gastrointestinais/química , Imunoglobulina A/sangue , Imunoglobulina G/sangue , Leucócitos/imunologia , Oligossacarídeos/administração & dosagem , Oligossacarídeos/química
10.
Nat Immunol ; 13(11): 1063-71, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23001144

RESUMO

The protein Tir (translocated intimin receptor) in enteric bacteria shares sequence similarity with the host cellular immunoreceptor tyrosine-based inhibition motifs (ITIMs). Despite the importance of Tir in pedestal formation, relatively little is known about the role of Tir and its ITIMs in the regulation of the host immune response. Here we demonstrate that Tir from enteropathogenic Escherichia coli (EPEC) interacted with the host cellular tyrosine phosphatase SHP-1 in an ITIM phosphorylation-dependent manner. The association of Tir with SHP-1 facilitated the recruitment of SHP-1 to the adaptor TRAF6 and inhibited the ubiquitination of TRAF6. Moreover, the ITIMs of Tir suppressed EPEC-stimulated expression of proinflammatory cytokines and inhibited intestinal immunity to infection with Citrobacter rodentium. Our findings identify a previously unknown mechanism by which bacterial ITIM-containing proteins can inhibit innate immune responses.


Assuntos
Infecções por Enterobacteriaceae/imunologia , Proteínas de Escherichia coli/imunologia , Motivo de Inibição do Imunorreceptor Baseado em Tirosina/imunologia , Intestinos/imunologia , Receptores de Superfície Celular/imunologia , Transdução de Sinais/imunologia , Animais , Células Cultivadas , Citrobacter rodentium/imunologia , Infecções por Enterobacteriaceae/genética , Infecções por Enterobacteriaceae/microbiologia , Escherichia coli Enteropatogênica/imunologia , Escherichia coli Enteropatogênica/patogenicidade , Proteínas de Escherichia coli/genética , Regulação da Expressão Gênica/imunologia , Interações Hospedeiro-Patógeno , Imunidade Inata , Imunidade nas Mucosas , Motivo de Inibição do Imunorreceptor Baseado em Tirosina/genética , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Intestinos/microbiologia , Macrófagos/imunologia , Macrófagos/microbiologia , Camundongos , Fosforilação , Ligação Proteica , Proteína Tirosina Fosfatase não Receptora Tipo 6/genética , Proteína Tirosina Fosfatase não Receptora Tipo 6/imunologia , Receptores de Superfície Celular/genética , Transdução de Sinais/genética , Fator 6 Associado a Receptor de TNF/genética , Fator 6 Associado a Receptor de TNF/imunologia , Ubiquitina/genética , Ubiquitina/imunologia , Ubiquitinação
11.
Vaccine ; 30(41): 5999-6005, 2012 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-22828590

RESUMO

Enteropathogenic Escherichia coli (EPEC) is an important cause of diarrhea in children. EPEC adheres to the intestinal epithelium and causes attaching and effacing (A/E) lesions. Recombinant Mycobacterium smegmatis (Smeg) and Mycobacterium bovis BCG strains were constructed to express either BfpA or intimin. The entire bfpA gene and a portion of the intimin gene were amplified by PCR from EPEC genomic DNA and inserted into the pMIP12 vector at the BamHI/KpnI sites. The pMIP_bfpA and pMIP_intimin vectors were introduced separately into Smeg and BCG. Recombinant clones were selected based on kanamycin resistance and designated rSmeg_pMIP_(bfpA or intimin) and rBCG_pMIP_(bfpA or intimin). The expression of bfpA and intimin was detected by Immunoblotting using polyclonal anti-BfpA and anti-intimin antibodies. The immunogenicity of these proteins was assessed in C57BL/6 mice by assaying the feces and serum for the presence of anti-BfpA and anti-intimin IgA and IgG antibodies. TNF-α and INF-γ were produced in vitro by spleen cells from mice immunized with recombinant BfpA, whereas TNF-γ was produced in mice immunized with recombinant intimin. The adhesion of EPEC (E2348/69) to HEp-2 target cells was blocked by IgA or IgG antibodies from mice immunized with recombinant BfpA or intimin but not by antibodies from non-immunized mice. Immunogenic non-infectious vectors containing relevant EPEC virulence genes may be promising vaccine candidates.


Assuntos
Adesinas Bacterianas/imunologia , Vacina BCG/administração & dosagem , Proteínas de Escherichia coli/imunologia , Proteínas de Fímbrias/imunologia , Mycobacterium smegmatis/imunologia , Adesinas Bacterianas/genética , Animais , Anticorpos Antibacterianos/sangue , Vacina BCG/imunologia , Linhagem Celular , Escherichia coli Enteropatogênica/imunologia , Escherichia coli Enteropatogênica/patogenicidade , Células Epiteliais/citologia , Células Epiteliais/microbiologia , Proteínas de Escherichia coli/genética , Feminino , Proteínas de Fímbrias/genética , Vetores Genéticos , Humanos , Interferon gama/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Mycobacterium bovis/genética , Mycobacterium bovis/imunologia , Mycobacterium smegmatis/genética , Baço/citologia , Baço/imunologia , Fator de Necrose Tumoral alfa/imunologia , Vacinas Sintéticas/administração & dosagem , Vacinas Sintéticas/imunologia
12.
Curr Biol ; 22(7): 627-31, 2012 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-22386311

RESUMO

The continuous monolayer of intestinal epithelial cells (IECs) lining the gut lumen functions as the site of nutrient absorption and as a physical barrier to prevent the translocation of microbes and associated toxic compounds into the peripheral vasculature. IECs also express host defense proteins such as intestinal alkaline phosphatase (IAP), which detoxify bacterial products and prevent intestinal inflammation. Our laboratory recently showed that IAP is enriched on vesicles that are released from the tips of IEC microvilli and accumulate in the intestinal lumen. Here, we show that these native "lumenal vesicles" (LVs) (1) contain catalytically active IAP that can dephosphorylate lipopolysaccharide (LPS), (2) cluster on the surface of native lumenal bacteria, (3) prevent the adherence of enteropathogenic E. coli (EPEC) to epithelial monolayers, and (4) limit bacterial population growth. We also find that IECs upregulate LV production in response to EPEC and other Gram-negative pathogens. Together, these results suggest that microvillar vesicle shedding represents a novel mechanism for distributing host defense machinery into the intestinal lumen and that microvillus-derived LVs modulate epithelial-microbial interactions.


Assuntos
Fosfatase Alcalina/metabolismo , Vesículas Citoplasmáticas/metabolismo , Escherichia coli Enteropatogênica/imunologia , Células Epiteliais/imunologia , Lipopolissacarídeos/metabolismo , Microvilosidades/metabolismo , Animais , Células CACO-2 , Vesículas Citoplasmáticas/microbiologia , Vesículas Citoplasmáticas/ultraestrutura , Enterócitos/citologia , Enterócitos/metabolismo , Escherichia coli Enteropatogênica/crescimento & desenvolvimento , Escherichia coli Enteropatogênica/metabolismo , Humanos , Intestino Delgado/citologia , Intestino Delgado/metabolismo , Intestino Delgado/microbiologia , Microscopia Eletrônica de Transmissão , Microvilosidades/microbiologia , Microvilosidades/ultraestrutura , Cadeias Pesadas de Miosina/metabolismo , Miosina Tipo I/metabolismo , Ratos
13.
PLoS One ; 6(10): e27030, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22046438

RESUMO

BACKGROUND: Enteropathogenic (EPEC) and Enteroaggregative (EAEC) E. coli have similar, but distinct clinical symptoms and modes of pathogenesis. Nevertheless when they infect the gastrointestinal tract, it is thought that their flagellin causes IL-8 release leading to neutrophil recruitment and gastroenteritis. However, this may not be the whole story as the effect of bacterial adherence to IEC innate response(s) remains unclear. Therefore, we have characterized which bacterial motifs contribute to the innate epithelial response to EPEC and EAEC, using a range of EPEC and EAEC isogenic mutant strains. METHODOLOGY: Caco-2 and HEp-2 cell lines were exposed to prototypical EPEC strain E2348/69 or EAEC strain O42, in addition to a range of isogenic mutant strains. E69 [LPS, non-motile, non-adherent, type three secretion system (TTSS) negative, signalling negative] or O42 [non-motile, non-adherent]. IL-8 and CCL20 protein secretion was measured. Bacterial surface structures were assessed by negative staining Transmission Electron Microscopy. The Fluorescent-actin staining test was carried out to determine bacterial adherence. RESULTS: Previous studies have reported a balance between the host pro-inflammatory response and microbial suppression of this response. In our system an overall balance towards the host pro-inflammatory response is seen with the E69 WT and to a greater extent O42 WT, which is in fit with clinical symptoms. On removal of the external EPEC structures flagella, LPS, BFP, EspA and EspC; and EAEC flagella and AAF, the host inflammatory response is reduced. However, removal of E69 lymphostatin increases the host inflammatory response suggesting involvement in the bacterial mediated anti-inflammatory response. CONCLUSION: Epithelial responses were due to combinations of bacterial agonists, with host-bacterial contact a key determinant of these innate responses. Host epithelial recognition was offset by the microbe's ability to down-regulate the inflammatory response. Understanding the complexity of this host-microbial balance will contribute to improved vaccine design for infectious gastroenteritis.


Assuntos
Aderência Bacteriana/imunologia , Escherichia coli Enteropatogênica/imunologia , Células Epiteliais/imunologia , Escherichia coli/imunologia , Imunidade Inata , Animais , Linhagem Celular , Interações Hospedeiro-Patógeno/imunologia , Inflamação/microbiologia
14.
Scand J Immunol ; 73(4): 268-83, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21204905

RESUMO

Enteropathogenic Escherichia coli (EPEC) causes diarrhoeal disease by altering enterocyte physiology and producing mucosal inflammation. Many details concerning the host response against EPEC remain unknown. We evaluated the role of EPEC virulence factors on the inflammatory response through an analysis of bacterial recognition, cell signalling, and cytokine production using an in vitro epithelial cell infection model. Interestingly, we found that EPEC infection recruits Toll-like receptor 5 (TLR5) to the cell surface. We confirmed that type 3 secretion system (T3SS) and flagellin (FliC) are necessary for efficient extracellular regulated kinases 1 and 2 (ERK1/2) activation and found that intimin could down-regulate this pathway. Besides flagellin, intimin was required to keep nuclear factor kappa B (NF-κB) activated during infection. EPEC infection activated tumour necrosis factor alpha (TNF-α) production and induced interleukin (IL)-1ß and IL-8 release. Virulence factors such as intimin, T3SS, EspA and fliC were required for IL-1ß secretion, whereas intimin and T3SS participated in IL-8 release. Flagellin was essential for late secretion of TNF-α and IL-8 and intimin stimulated cytokine secretion. Initial adherence limited TNF-α release, whereas late attachment sustained TNF-α secretion. We conclude that intimin modulates TLR5 activation and intimate adherence alters the proinflammatory response.


Assuntos
Aderência Bacteriana/imunologia , Escherichia coli Enteropatogênica/imunologia , Células Epiteliais/imunologia , Células Epiteliais/microbiologia , Intestinos/citologia , Receptor 5 Toll-Like/metabolismo , Transporte Ativo do Núcleo Celular/fisiologia , Adesinas Bacterianas/fisiologia , Sistemas de Secreção Bacterianos/fisiologia , Membrana Celular/metabolismo , Escherichia coli Enteropatogênica/genética , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Proteínas de Escherichia coli/fisiologia , Flagelina/genética , Flagelina/metabolismo , Expressão Gênica/genética , Expressão Gênica/imunologia , Técnicas de Inativação de Genes , Células HT29 , Humanos , Proteínas I-kappa B/metabolismo , Inflamação/imunologia , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Interleucina-8/genética , Interleucina-8/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Inibidor de NF-kappaB alfa , NF-kappa B/metabolismo , Fosforilação/fisiologia , Transporte Proteico , Receptor 4 Toll-Like/metabolismo , Receptor 5 Toll-Like/genética , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo , Fatores de Virulência/fisiologia
15.
EMBO J ; 30(1): 221-31, 2011 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-21113130

RESUMO

Two major arms of the inflammatory response are the NF-κB and c-Jun N-terminal kinase (JNK) pathways. Here, we show that enteropathogenic Escherichia coli (EPEC) employs the type III secretion system to target these two signalling arms by injecting host cells with two effector proteins, NleC and NleD. We provide evidence that NleC and NleD are Zn-dependent endopeptidases that specifically clip and inactivate RelA (p65) and JNK, respectively, thus blocking NF-κB and AP-1 activation. We show that NleC and NleD co-operate and complement other EPEC effectors in accomplishing maximal inhibition of IL-8 secretion. This is a remarkable example of a pathogen using multiple effectors to manipulate systematically the host inflammatory response signalling network.


Assuntos
Escherichia coli Enteropatogênica/fisiologia , Infecções por Escherichia coli/imunologia , Proteínas de Escherichia coli/imunologia , Interações Hospedeiro-Patógeno , Proteínas Quinases JNK Ativadas por Mitógeno/imunologia , NF-kappa B/imunologia , Apoptose , Escherichia coli Enteropatogênica/imunologia , Proteínas de Escherichia coli/genética , Expressão Gênica , Células HeLa , Humanos , Interleucina-8/genética , Interleucina-8/imunologia , Proteína Quinase 9 Ativada por Mitógeno/imunologia , Fator de Transcrição RelA/imunologia , Transcrição Gênica , Fator de Necrose Tumoral alfa/imunologia
16.
Microbiology (Reading) ; 155(Pt 10): 3214-3225, 2009 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19628559

RESUMO

Enterohaemorrhagic and enteropathogenic Escherichia coli (EHEC and EPEC) inject a repertoire of effector proteins into host cells via a type III secretion system (T3SS) encoded by the locus of enterocyte effacement (LEE). OspG is an effector protein initially identified in Shigella that was shown to inhibit the host innate immune response. In this study, we found ospG homologues in EHEC (mainly of serogroup O111) and in Yersinia enterocolitica. The T3SS encoded by the LEE was able to inject these different OspG homologues into host cells. Infection of HeLa cells with EHEC O111 inhibited the NF-kappaB-dependent innate immune response via a T3SS-dependent mechanism. However, an EHEC O111 ospG mutant was still able to inhibit NF-kappaB p65 transfer to the nucleus in infected cells stimulated by tumour necrosis factor alpha (TNF-alpha). In addition, no difference in the inflammatory response was observed between wild-type EHEC O111 and the isogenic ospG mutant in the rabbit ligated intestinal loop model. These results suggest that OspG is not the sole effector protein involved in the inactivation of the host innate immune system during EHEC O111 infection.


Assuntos
Escherichia coli Êntero-Hemorrágica/imunologia , Escherichia coli Êntero-Hemorrágica/patogenicidade , Infecções por Escherichia coli/imunologia , Infecções por Escherichia coli/microbiologia , Proteínas de Escherichia coli/fisiologia , Imunidade Inata , NF-kappa B/antagonistas & inibidores , Fatores de Virulência/fisiologia , Sequência de Aminoácidos , Animais , Escherichia coli Enteropatogênica/imunologia , Escherichia coli Enteropatogênica/patogenicidade , Infecções por Escherichia coli/patologia , Ordem dos Genes , Células HeLa , Humanos , Intestinos/microbiologia , Intestinos/patologia , Dados de Sequência Molecular , Coelhos , Alinhamento de Sequência , Sorotipagem
17.
São Paulo; s.n; 16 abr. 2009. 134 p. graf, tab, ilus.
Tese em Português | LILACS | ID: lil-525239

RESUMO

A diarréia é um importante problema de saúde pública no mundo inteiro e a Escherichía coli é um dos mais freqüentes microorganismos causadores desta doença. A Escherichia coli enteropatogênica (EPEC), um dos principais agentes etiológicos das diarréias infantis no nosso país, é genética e fenotipicamente relacionada com a E. colí enterohemorrágica (EHEC) que além de provocar diarréia é responsável por complicações como síndrome hemolítica urêmica (HUS) e colite hemorrágica (HC). Embora a EHEC seja considerada emergente pela OMS, no Brasil poucos casos de complicações como HUS e HC foram reportados. O mecanismo de patogenicidade comum entre EPEC e EHEC é conhecido como a lesão "attaching and effacing" nos microvilos do enterócito. Esta lesão é mediada por um conjunto de fatores de virulência, dentre eles a intimina. A intimina é uma proteína de membrana externa, responsável pelo íntimo contato da bactéria com o enterócito, possui uma região N-terminal que é altamente conservada e uma região C-terminal que é variável. De acordo com a região variável, existem vários subtipos de intimina, dentre eles as intiminas , α, β e γ...


Assuntos
Humanos , Masculino , Feminino , Adulto , Anticorpos/genética , Anticorpos/imunologia , Diarreia/genética , Diarreia/imunologia , Escherichia coli Enteropatogênica/fisiologia , Escherichia coli Enteropatogênica/imunologia , Fatores de Virulência/genética , Fatores de Virulência/imunologia , Infecções por Escherichia coli/imunologia , Colostro , Ensaio de Imunoadsorção Enzimática , Métodos Analíticos de Preparação de Amostras , Soro , Interpretação Estatística de Dados
18.
Cell Microbiol ; 11(3): 521-30, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19134113

RESUMO

In vitro organ culture (IVOC) represents a gold standard model to study enteropathogenic E. coli (EPEC) infection of human intestinal mucosa. However, the optimal examination of the bacterial-host cell interaction requires a directional epithelial exposure, without serosal or cut surface stimulation. A polarized IVOC system (pIVOC) was developed in order to overcome such limitations: apical EPEC infection produced negligible bacterial leakage via biopsy edges, resulted in enhanced colonization compared with standard IVOC, and showed evidence of bacterial detachment, as in natural rabbit EPEC infections. Examination of mucosal innate immune responses in pIVOC showed both interleukin (IL)-8 mRNA and protein levels were significantly increased after apical EPEC infection. Increased IL-8 levels mainly depended on flagellin expression as fliC-negative EPEC did not elicit a significant IL-8 response despite increased mucosal colonization compared with wild-type EPEC. In addition, apical application of purified flagella significantly increased IL-8 protein levels over non-infected controls. Immunofluorescence staining of EPEC-infected small intestinal biopsies revealed apical and basolateral distribution of Toll-like receptor (TLR) 5 on epithelium, suggesting that EPEC can trigger mucosal IL-8 responses by apical flagellin/TLR5 interaction ex vivo and does not require access to the basolateral membrane as postulated in cell culture models.


Assuntos
Escherichia coli Enteropatogênica/crescimento & desenvolvimento , Mucosa Intestinal/microbiologia , Escherichia coli Enteropatogênica/imunologia , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/imunologia , Flagelina , Deleção de Genes , Perfilação da Expressão Gênica , Humanos , Interleucina-8/biossíntese , Mucosa Intestinal/patologia , Técnicas de Cultura de Órgãos/métodos , Receptor 5 Toll-Like/biossíntese
19.
Int J Med Microbiol ; 299(2): 121-32, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18771950

RESUMO

Enteropathogenic Escherichia coli cause protracted diarrhoea and malnutrition in infants by cytoskeletal depolymerisation and effacement of enterocyte microvilli. In this study, outer membrane proteins of wild-type enteropathogenic E. coli and an intimin-deficient mutant are shown to induce apoptosis by up-regulation of tumour necrosis factor alpha and activation of c-jun N-terminal kinase. Fluorescence-activated cell sorter analysis revealed apoptosis of cells treated with outer membrane proteins of wild-type and intimin-deficient strains. Proteinase K treatment of outer membrane proteins reduced apoptosis significantly, as did neutralising tumour necrosis factor alpha with specific antibodies. Elevated tumour necrosis factor receptor 1-associated death domain and caspase-3 expression were also observed on treatment with both types of outer membrane proteins. Furthermore, apoptosis was associated with suppression of Bcl-2 protein expression, up-regulation of Bax mRNA levels and increased cytochrome c release from mitochondria. Elevated phospho-c-jun N-terminal kinase, c-jun mRNA and activator protein-1 expression were observed, and phosphorylation of activator protein-1 was also observed by DNA-binding assays. Inhibition of c-jun N-terminal kinase, but not inhibition of p38 mitogen-activated protein kinase, resulted in reduction of tumour necrosis factor alpha mRNA levels and caspase-3 protein levels, and a reduction in apoptosis as observed by fluorescence-activated cell sorter analysis. From the host point of view, this study suggests a possible interplay between the death receptor and mitochondrial pathways when cell-free bacterial outer membrane preparations are used to trigger apoptosis.


Assuntos
Apoptose , Proteínas da Membrana Bacteriana Externa/imunologia , Escherichia coli Enteropatogênica/imunologia , Células Epiteliais/microbiologia , Caspase 3/biossíntese , Linhagem Celular Tumoral , Citocromos c/metabolismo , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/biossíntese , Proteínas Proto-Oncogênicas c-bcl-2/antagonistas & inibidores , Fator de Necrose Tumoral alfa/biossíntese , Regulação para Cima , Proteína X Associada a bcl-2/biossíntese
20.
Biochemistry ; 47(44): 11590-7, 2008 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-18842005

RESUMO

Escherichia coli O86 possesses high human blood group B activity because of its O-antigen structure, sharing the human blood group B epitope. In this study, the wbwK gene of E. coli O86:B7 was expressed and purified as the GST fusion protein. Thereafter, the wbwK gene was biochemically identified to encode an alpha1,2-fucosyltransferase through radioactivity assays, as well as mass spectrometry and NMR spectroscopy. WbwK shows strict substrate specificity and only recognizes Gal beta1,3GalNAc alpha-OR (T-antigen and derivatives) as the acceptor to generate the H-type 3 blood group antigen. In contrast to other alpha1,2-fucosyltransferases, WbwK does not display activity toward the simple substrate Gal beta-OMe. Comparison with another recently characterized alpha1,2-fucosyltransferase (WbsJ) of E. coli O128:B12 indicates a low level of amino acid identity between them; however, they share a common acceptor substrate, Gal beta1,3GalNAc alpha-OR. Domain swapping between WbwK and WbsJ revealed that the smaller variable domains located in the C-terminus determine substrate specificity, whereas the larger variable domain in the N-terminus might play a role in forming the correct conformation for substrate binding or for localization of the alpha1,2-fucosyltransferase involved in O-antigen biosynthesis. In addition, milligram scale biosynthesis of the H-type 3 blood group antigen was explored using purified recombinant WbwK. WbwK may have potential applications in masking T-antigen, the tumor antigen, in vivo.


Assuntos
Antígenos de Bactérias/biossíntese , Escherichia coli Enteropatogênica/enzimologia , Escherichia coli Enteropatogênica/imunologia , Fucosiltransferases/metabolismo , Antígenos O/biossíntese , Sequência de Aminoácidos , Antígenos de Bactérias/química , Sequência de Carboidratos , Escherichia coli Enteropatogênica/genética , Fucosiltransferases/genética , Genes Bacterianos , Humanos , Cinética , Dados de Sequência Molecular , Antígenos O/química , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Homologia de Sequência de Aminoácidos , Especificidade por Substrato , Galactosídeo 2-alfa-L-Fucosiltransferase
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA